You are on page 1of 5

FOCUS

Cite as: M. Bosmann, Sci. Immunol.


10.1126/sciimmunol.abj1014 (2021).

CORONAVIRUS

Complement control for COVID-19


Markus Bosmann1,2,3,4*
1
Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, 02118, MA, USA. 2Center for Thrombosis and Hemostasis, University Medical
Center Mainz, 55131 Mainz, Germany. 3Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, 02118, USA. 4Research Center
for Immunotherapy (FZI), University Medical Center Mainz, 55131 Mainz, Germany.
*Corresponding author. Email: mbosmann@bu.edu

Excessive complement activation contributes to lung disease and adverse patient outcomes in COVID-19
(see the related Research Articles by Yan et al. and Ma et al.).

Downloaded from http://immunology.sciencemag.org/ by guest on May 26, 2021


The complement system is an integral part of innate immune anaphylatoxin C5a. C5b is the starting point of the pore-
defense. It consists of about 50 proteins in plasma, on cell forming membrane attack complex (MAC) consisting of C5b-
surfaces and inside host cells. The traditional view is that C9 with a channel diameter of ~100 Å. The C3/C5 hub
complement proteins guard the local extracellular spaces and represents a gigantic amplification loop. The alternative
systemic bloodstream against invading pathogens. Loss-of- C3bBb convertase (half-life ~3 min) cleaves additional C3,
function mutations resulting in terminal complement resulting in more C3bBb and so on and so forth. This
pathway deficiencies are associated with a 10,000-fold higher enzymatic chain reaction can deposit millions of C3b
risk for life-threatening meningococcal infections in humans. molecules on target surfaces in a few seconds. It is no surprise
Surprisingly, the complement system is redundant for that such explosive events need to be tightly regulated to
defense against most pathogens except encapsulated maintain the delicate balance of effective and justified
bacteria. Newer concepts embrace the view that complement pathogen attack, while avoiding damage of innocent
factors mediate functions inside cells either directly or bystander cells.
through surface receptors. Complement activity fine-tunes In the April and May issues of Science Immunology, two
homeostasis, metabolism and biogenesis. On the other hand, articles highlight the importance of complement activation
uncontrolled complement activation causes disease and can in severe COVID-19. Ma et al. report an observational clinical
even worsen the outcome of infections. Toxic complement study of a total of n=259 participants classified in subcohorts
effectors mediate tissue destruction and organ injury during of critically ill SARS-CoV-2 patients (from two clinical cen-
inflammatory diseases. Acute respiratory distress syndrome ters) compared to patients with influenza or acute respiratory
(ARDS) and sepsis are frequent and severe complications of failure of other causes requiring invasive mechanical ventila-
acute infections and notorious for excessive complement tion (1). Higher complement activation products in blood cor-
consumption. The three pathways of complement activation related with and predicted increased disease severity. The
are designed for immune sensing of non-self surfaces and concentrations of soluble MAC (sC5b-C9) were higher
foreign antigens. The mannose-binding lectin (MBL)/ficolin (p<0.0001) in COVID-19 patients as compared to influenza
pathway starts with soluble pathogen pattern recognition patients. SARS-CoV-2-associated acute respiratory failure
receptors as sensors for foreign carbohydrate motifs (Fig. 1). showed increased sC5b-C9 compared to ARDS from non-
The alternative pathway is fueled by a spontaneous COVID-19 etiologies. The plasma levels of sC5b-C9 and C5a
‘smoldering’ hydrolysis of C3 targeting all surfaces, unless correlated as expected, since both analytes arise together
these surfaces present complement inhibitory proteins from C5 cleavage, although C5a has an ultra-short plasma
(CD46, CD55, CD59) as a protective self-signal. This C3 ‘tick- half-life (<5 min). C5a is rapidly inactivated by plasma car-
over’ is sustained by the high concentrations of C3 in plasma boxypeptidase N and B, which remove the C-terminal argi-
(1-2 g/L) – the highest level of all complement factors. The nine residue from the polypeptide chain. The degradation
classical pathway is initiated by antigen-antibody complexes product C5adesArg has low affinity for the C5aR1 receptor but
which are recognized by the multimeric C1 complex. As a retains binding to the homologous C5aR2 receptor. Comple-
safeguard, IgG antibodies bound in clusters or pentameric ment diagnostics remains a technically challenging field, and
IgM are required to surpass the activation threshold. All not just because of the volatile nature of activation products.
complement pathways converge on C3 convertase complexes Antibody-based tests must selectively recognize the ne-
leading to C3 cleavage into the larger C3b and the smaller oepitopes of complement proteins which are created by pro-
anaphylactic C3a peptides. C3b is essential for the formation teolytic cleavage. Cross-reactivity of anti-C5a antibodies with
of C5 convertase for cleavage of C5 into C5b and the C5 or C5b can be a serious impediment.

First release: 25 May 2021 immunology.sciencemag.org (Page numbers not final at time of first release) 1
Ma et al. also addressed which of the three complement increased by SARS-CoV-2 pseudovirus infection, primes S-
activation pathways is engaged (1). The authors found higher protein and enhances virus entry (8). In addition, invading
concentrations of Factor B and Ba in severe COVID-19 pa- pathogens can carry bound C3 into the cytosol (9). The intra-
tients who required intensive care treatment and in non-sur- cellular C3 flags viruses for proteasomal degradation and
vivors. Factor B is a zymogen in the alternative pathway. contributes to the activation of the viral RNA-sensing MAVS
SARS-CoV-2 infection down-regulates expression of CD46, signaling pathway (9). Interestingly, several viruses have
CD55 and CD59 (2), which may contribute to increased alter- evolved evasion strategies to cleave C3 with viral proteases
native pathway activation. It is worth mentioning that the (9).
C3bBb amplification loop is also triggered by the MBL/ficolin Yan et al. further characterize that SARS-CoV-2-induced
and classical pathways. The SARS-CoV-2 S-protein contains expression of C3 and Factor B was dependent on type I inter-
numerous N-acetylglucosamine moieties (3). N-acetylglu- ferons, the interferon-activated JAK1/2-STAT1 signaling
cosamine is recognized by ficolins. SARS-CoV-2 N-protein is pathway and NF-κB RelA (6). The JAK1/2 inhibitor rux-
likewise glycosylated and directly binds to MASP2 of the olitinib reduced generation of C3a, presumably by shutting

Downloaded from http://immunology.sciencemag.org/ by guest on May 26, 2021


MBL/ficolin pathway. The classical pathway may be activated off C3 and Factor B production during SARS-CoV-2 infection.
in the later phases of infection by virus-specific antibodies In conclusion, complement activation is a predominant
(Fig. 1) and COVID-19-associated autoantibodies. The latter feature of severe COVID-19. Several approaches are conceiv-
could fuel tissue injury. Lastly, Ma et al. describe the statisti- able to control complement activation as a therapeutic prin-
cal correlation of Factor D (which cleaves Factor B in the al- ciple in COVID-19 (9). A number of complement blockers are
ternative pathway) with markers of endothelial cell injury either readily available for drug repurposing or in advanced
(Ang2) and coagulation such as von Willebrand Factor (vWF) stages of drug development (Fig. 1). Eculizumab is a mono-
which is released by activated platelets and endothelial cells clonal antibody to block C5 conversion and the first drug of
(1). The cross-talk between coagulation and complement its class. Eculizumab improves the life expectancy of patients
could contribute to COVID-19-associated coagulopathy and with paroxysmal nocturnal hemoglobinuria (an acquired de-
thromboembolic events. Thrombin can directly cleave C5 to ficiency in glycosylphosphatidylinositol-anchored CD55 and
bypass C3 (4). C5a ligation with C5aR1 and C5aR2 induces CD59) and is effective in treating atypical hemolytic uremic
extracellular trap formation and appearance of externalized syndrome (aHUS) and neuromyelitis optica spectrum disor-
histones (5), which are procoagulant and provoke lung injury. der (NMOSD). C3 inhibition can be achieved by AMY-101 and
In the second article, Yan et al. identify that the tran- pegcetacoplan. It is too early to predict which complement
scriptomic signatures of SARS-CoV-2 infected human lung blocker may have the best efficacy and a favorable risk profile
epithelial cells were enriched for changes in complement to selectively suppress hyperinflammation while sparing the
gene expression (6). SARS-CoV-2 induced Factor B and C3 as protective antiviral activities of complement. Clinical trials to
the central players of the alternative pathway, but also up- control complement activation and improve patient out-
regulated other complement genes such as C1r and C1s. The comes of COVID-19 are underway.
expression levels of C3 correlated with viral loads. In scRNA- REFERENCES AND NOTES
Seq data sets from biopsies and lavages of COVID-19 patients, 1. L. Ma, S. K. Sahu, M. Cano, V. Kuppuswamy, J. Bajwa, J. McPhatter, A. Pine, M.
C3 was up-regulated in type II alveolar epithelial cells (AT2s) Meizlish, G. Goshua, C.-H. Chang, H. Zhang, C. Price, P. Bahel, H. Rinder, T. Lei, A.
and the C3aR receptor was induced in macrophages and Day, D. Reynolds, X. Wu, R. Schriefer, A. M. Rauseo, C. W. Goss, J. A. O’Halloran,
R. M. Presti, A. H. Kim, A. E. Gelman, C. D. Cruz, A. I. Lee, P. Mudd, H. J. Chun, J. P.
monocytes. It is appreciated that extrahepatic production of Atkinson, H. S. Kulkarni, Increased complement activation is a distinctive feature
complement factors occurs at proximal sites of pathogen en- of severe SARS-CoV-2 infection. Sci. Immunol. 6, eabh2259 (2021).
counters such as in organs with mucosal surfaces and in leu- doi:10.1126/sciimmunol.abh2259 Medline
kocytes. However, not all lung cell types produce complement 2. J. Huang, A. J. Hume, K. M. Abo, R. B. Werder, C. Villacorta-Martin, K.-D.
Alysandratos, M. L. Beermann, C. Simone-Roach, J. Lindstrom-Vautrin, J. Olejnik,
factors and no single cell type expresses all complement E. L. Suder, E. Bullitt, A. Hinds, A. Sharma, M. Bosmann, R. Wang, F. Hawkins, E. J.
genes in relevant quantities. Yan et al. found that C3 protein Burks, M. Saeed, A. A. Wilson, E. Mühlberger, D. N. Kotton, SARS-CoV-2 infection
was processed to its active form in SARS-CoV-2 infected lung of pluripotent stem cell-derived human lung alveolar type 2 cells elicits a rapid
epithelial cells (Calu-3 and iPSC-derived AT2s). C3a was de- epithelial-intrinsic inflammatory response. Cell Stem Cell 27, 962–973.e7 (2020).
doi:10.1016/j.stem.2020.09.013 Medline
tected in the lung epithelial cells and existed in direct linear 3. R. Wintjens, A. M. Bifani, P. Bifani, Impact of glycan cloud on the B-cell epitope
correlation with SARS-CoV-2 N-protein. The C3 activation prediction of SARS-CoV-2 Spike protein. NPJ Vaccines 5, 81 (2020).
was prevented by a cell-permeable Factor B inhibitor. The au- doi:10.1038/s41541-020-00237-9 Medline
thors interpret their findings in support of the concept of a 4. M. Huber-Lang, J. V. Sarma, F. S. Zetoune, D. Rittirsch, T. A. Neff, S. R. McGuire, J.
D. Lambris, R. L. Warner, M. A. Flierl, L. M. Hoesel, F. Gebhard, J. G. Younger, S. M.
non-traditional intracellular complement activation mecha- Drouin, R. A. Wetsel, P. A. Ward, Generation of C5a in the absence of C3: A new
nism. Cathepsin L can process C3 into biologically active C3a complement activation pathway. Nat. Med. 12, 682–687 (2006).
and C3b (7). Interestingly, Cathepsin L expression is doi:10.1038/nm1419 Medline

First release: 25 May 2021 immunology.sciencemag.org (Page numbers not final at time of first release) 2
5. M. Bosmann, J. J. Grailer, R. Ruemmler, N. F. Russkamp, F. S. Zetoune, J. V. Sarma,
T. J. Standiford, P. A. Ward, Extracellular histones are essential effectors of C5aR-
and C5L2-mediated tissue damage and inflammation in acute lung injury. FASEB
J. 27, 5010–5021 (2013). doi:10.1096/fj.13-236380 Medline
6. B. Yan, T. Freiwald, D. Chauss, L. Wang, E. West, C. Mirabelli, C. J. Zhang, E.-M.
Nichols, N. Malik, R. Gregory, M. Bantscheff, S. Ghidelli-Disse, M. Kolev, T. Frum,
J. R. Spence, J. Z. Sexton, K. D. Alysandratos, D. N. Kotton, S. Pittaluga, J. Bibby,
N. Niyonzima, M. R. Olson, S. Kordasti, D. Portilla, C. E. Wobus, A. Laurence, M. S.
Lionakis, C. Kemper, B. Afzali, M. Kazemian, SARS-CoV-2 drives JAK1/2-
dependent local complement hyperactivation. Sci. Immunol. 6, eabg0833 (2021).
doi:10.1126/sciimmunol.abg0833 Medline
7. M. K. Liszewski, M. Kolev, G. Le Friec, M. Leung, P. G. Bertram, A. F. Fara, M. Subias,
M. C. Pickering, C. Drouet, S. Meri, T. P. Arstila, P. T. Pekkarinen, M. Ma, A. Cope,
T. Reinheckel, S. Rodriguez de Cordoba, B. Afzali, J. P. Atkinson, C. Kemper,
Intracellular complement activation sustains T cell homeostasis and mediates
effector differentiation. Immunity 39, 1143–1157 (2013).

Downloaded from http://immunology.sciencemag.org/ by guest on May 26, 2021


doi:10.1016/j.immuni.2013.10.018 Medline
8. M. M. Zhao, W.-L. Yang, F.-Y. Yang, L. Zhang, W.-J. Huang, W. Hou, C.-F. Fan, R.-H.
Jin, Y.-M. Feng, Y.-C. Wang, J.-K. Yang, Cathepsin L plays a key role in SARS-CoV-
2 infection in humans and humanized mice and is a promising target for new drug
development. Signal Transduct. Target. Ther. 6, 134 (2021). doi:10.1038/s41392-
021-00558-8 Medline
9. J. C. Tam, S. R. Bidgood, W. A. McEwan, L. C. James, Intracellular sensing of
complement C3 activates cell autonomous immunity. Science 345, 1256070
(2014). doi:10.1126/science.1256070 Medline
10. A. M. Risitano et al., Complement as a target in COVID-19? Nat. Rev. Immunol.
(2020).
Acknowledgments: The author thanks Catherine O’Neal for reading the manuscript,
Archana Jayaraman for assistance and the Evans Center for Interdisciplinary
Biomedical Research at Boston University School of Medicine for their support of
the Affinity Research Collaborative on ‘Respiratory Viruses: A Focus on COVID-
19’. Funding: The author’s research is supported by the National Institutes of
Health (1R01HL141513, 1R01HL139641, 1R01AI153613, 1UL1TR001430) and the
Deutsche Forschungsgemeinschaft (BO3482/3-3). Competing Interests: The
author declares no competing interests.

Published First Release 25 May 2021


10.1126/sciimmunol.abj1014

First release: 25 May 2021 immunology.sciencemag.org (Page numbers not final at time of first release) 3
Downloaded from http://immunology.sciencemag.org/ by guest on May 26, 2021
Fig. 1. Current concepts of complement activation and potential therapeutic interventions in severe COVID-19.
SARS-CoV-2 infection of alveolar epithelial cells (type II and type I) and subsequent interferon-dependent JAK1/2-
STAT1–induced expression of C3 and Factor B culminates in nontraditional intracellular processing of complement
proteins. In the extracellular spaces, SARS-CoV-2 activates complement via the mannose-binding lectin (MBL)/ficolin
pathway which senses glycosylated S- and N-proteins. The C3 ‘tick-over’ of the alternative pathway is accelerated by
a lack of complement inhibitors (CD46, CD55, CD59) on infected host cells and virions. The classical pathway is
activated by antibodies against viral antigens and COVID-19–associated autoantibodies. All complement activation
mechanisms converge on the C3/C5 hub to form the membrane-attack complex (MAC, C5b-C9) and generate
anaphylatoxins (C3a, C5a). These effectors recruit myeloid cells and cause endothelial activation, endothelial injury,
coagulopathy and hyperinflammation. The figure shows the molecular targets of several complement-inhibiting drugs
proposed as COVID-19 treatments: eculizumab and ravulizumab block C5 conversion, AMY-101 and pegcetacoplan
antagonize C3 activation, the IFX-1 monoclonal antibody inhibits C5a, and avdoralimab blocks the C5aR1 receptor.
Abbreviations: vWF: von Willebrand Factor, Ang2: Angiopoietin-2, C3aR: C3a receptor, C5aR1: C5a receptor 1, C5aR2:
C5a receptor 2, MASP1/2: Mannan-binding lectin serine proteases 1 and 2, ACE2: Angiotensin-converting enzyme 2,
TMPRSS2: Transmembrane protease, serine 2.
CREDIT: KELLIE HOLOSKI/SCIENCE IMMUNOLOGY
First release: 25 May 2021 immunology.sciencemag.org (Page numbers not final at time of first release) 4
Complement control for COVID-19
Markus Bosmann

Sci. Immunol. 6, eabj1014.


DOI: 10.1126/sciimmunol.abj1014

Downloaded from http://immunology.sciencemag.org/ by guest on May 26, 2021


ARTICLE TOOLS http://immunology.sciencemag.org/content/6/59/eabj1014

REFERENCES This article cites 9 articles, 3 of which you can access for free
http://immunology.sciencemag.org/content/6/59/eabj1014#BIBL

Use of this article is subject to the Terms of Service

Science Immunology (ISSN 2470-9468) is published by the American Association for the Advancement of Science, 1200 New
York Avenue NW, Washington, DC 20005. The title Science Immunology is a registered trademark of AAAS.
Copyright © 2021, American Association for the Advancement of Science

You might also like